Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
1.
Acta Pharmacol Sin ; 44(7): 1416-1428, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-36721007

RESUMEN

Non-alcoholic fatty liver disease (NAFLD) is a major health concern worldwide, and the incidence of metabolic disorders associated with NAFLD is rapidly increasing because of the obesity epidemic. There are currently no approved drugs that prevent or treat NAFLD. Recent evidence shows that bavachin, a flavonoid isolated from the seeds and fruits of Psoralea corylifolia L., increases the transcriptional activity of PPARγ and insulin sensitivity during preadipocyte differentiation, but the effect of bavachin on glucose and lipid metabolism remains unclear. In the current study we investigated the effects of bavachin on obesity-associated NAFLD in vivo and in vitro. In mouse primary hepatocytes and Huh7 cells, treatment with bavachin (20 µM) significantly suppressed PA/OA or high glucose/high insulin-induced increases in the expression of fatty acid synthesis-related genes and the number and size of lipid droplets. Furthermore, bavachin treatment markedly elevated the phosphorylation levels of AKT and GSK-3ß, improving the insulin signaling activity in the cells. In HFD-induced obese mice, administration of bavachin (30 mg/kg, i.p. every other day for 8 weeks) efficiently attenuated the increases in body weight, liver weight, blood glucose, and liver and serum triglyceride contents. Moreover, bavachin administration significantly alleviated hepatic inflammation and ameliorated HFD-induced glucose intolerance and insulin resistance. We demonstrated that bavachin protected against HFD-induced obesity by inducing fat thermogenesis and browning subcutaneous white adipose tissue (subWAT). We revealed that bavachin repressed the expression of lipid synthesis genes in the liver of obese mice, while promoting the expression of thermogenesis, browning, and mitochondrial respiration-related genes in subWAT and brown adipose tissue (BAT) in the mice. In conclusion, bavachin attenuates hepatic steatosis and obesity by repressing de novo lipogenesis, inducing fat thermogenesis and browning subWAT, suggesting that bavachin is a potential drug for NAFLD therapy.


Asunto(s)
Resistencia a la Insulina , Enfermedad del Hígado Graso no Alcohólico , Animales , Ratones , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Ratones Obesos , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Hígado/metabolismo , Obesidad/complicaciones , Obesidad/tratamiento farmacológico , Obesidad/genética , Flavonoides/farmacología , Dieta , Glucosa/metabolismo , Insulina/metabolismo , Dieta Alta en Grasa , Ratones Endogámicos C57BL
2.
Can J Physiol Pharmacol ; 99(10): 1079-1087, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33915055

RESUMEN

Chronic cerebral hypoperfusion (CCH), as a critical factor of chronic cerebrovascular diseases, has greatly influenced the health of patients with vascular dementia. Vitexin, a flavone C-glycoside (apigenin-8-C-ß-D-glucopyranoside) that belongs to the flavone subclass of flavonoids, has been shown to possess antioxidant and anti-ischemic properties; however, the putative protective effects of vitexin on the CCH need further investigation. In the current study, the role of vitexin and its underlying mechanism were investigated with permanent bilateral common carotid artery occlusion (2VO) in rats as well as mouse hippocampal neuronal (HT22) cells with oxygen and glucose deprivation/reoxygenation (OGD/R) injury model. The results demonstrated that vitexin improved cognitive dysfunction as well as alleviated pathological neuronal damage in hematoxylin plus eosin (HE) and TUNEL results. The decreased levels of exchange protein directly activated by cAMP 1 (Epac1), Epac2, Ras-associated protein 1 (Rap1), and phospho-extracellular signal-regulated kinase (p-ERK) were reversed by vitexin in rats with CCH. Furthermore, this study indicated that vitexin alleviated CCH-induced inflammation injuries by reducing the expression of NOD-like receptor 3 (NLRP3), caspase-1, interleukin 1ß (IL-1ß), IL-6, and cleaved caspase-3. In vitro, vitexin increased the expression of Epac1 and Epac2, decreased the activation of the NLRP3-mediated inflammation, and improved cell viability. Taken together, our findings suggest that vitexin can reduce the degree of the progressing pathological damage in the cortex and hippocampus and inhibit further deterioration of cognitive function in rats with CCH. Epac and NLRP3 can be regulated by vitexin in vivo and in vitro, which provides enlightenment for the protection of CCH injury.


Asunto(s)
Apigenina/farmacología , Trastornos Cerebrovasculares/tratamiento farmacológico , Factores de Intercambio de Guanina Nucleótido/metabolismo , Hipocampo/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Neuronas/efectos de los fármacos , Animales , Circulación Cerebrovascular , Trastornos Cerebrovasculares/metabolismo , Trastornos Cerebrovasculares/patología , Enfermedad Crónica , Trastornos del Conocimiento/metabolismo , Trastornos del Conocimiento/patología , Trastornos del Conocimiento/prevención & control , Factores de Intercambio de Guanina Nucleótido/genética , Hipocampo/metabolismo , Hipocampo/patología , Masculino , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Neuronas/metabolismo , Neuronas/patología , Ratas , Ratas Sprague-Dawley
3.
Cell Biol Int ; 44(7): 1481-1490, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32181963

RESUMEN

Pyroptosis is a form of cell death that is uniquely dependent on caspase-1. Pyroptosis involved in oxidized low-density lipoprotein (ox-LDL)-induced human macrophage death through the promotion of caspase-1 activation is important for the formation of unstable plaques in atherosclerosis. The mitochondrial outer membrane protein NIX directly interacts with microtubule-associated protein 1 light chain 3 (LC3). Although we previously showed that NIX-mediated mitochondrial autophagy is involved in the clearance of damaged mitochondria, how NIX contributes to ox-LDL-induced macrophage pyroptosis remains unknown. Here, immunoperoxidase staining Nix expression decreased in human atherosclerosis. When we silenced NIX expression in murine macrophage cell, active caspase-1, and mature interleukin-1ß expression levels were increased and LC3 was reduced. In addition, LDH release and acridine orange and ethidium bromide staining indicated that damage to macrophage cell membranes induced by ox-LDL was substantially worse. Moreover, intracellular reactive oxygen species and NLRP3 inflammasome levels increased. Taken together, these results demonstrated that NIX inhibits ox-LDL-induced macrophage pyroptosis via autophagy in atherosclerosis.


Asunto(s)
Aterosclerosis/patología , Autofagia/efectos de los fármacos , Lipoproteínas LDL/farmacología , Macrófagos/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Aterosclerosis/inducido químicamente , Aterosclerosis/metabolismo , Muerte Celular/efectos de los fármacos , Humanos , Inflamasomas/efectos de los fármacos , Inflamasomas/metabolismo , Lipoproteínas LDL/metabolismo , Macrófagos/metabolismo , Macrófagos/patología , Mitocondrias/metabolismo
4.
Exp Mol Pathol ; 114: 104402, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32061942

RESUMEN

The aim of this study was to investigate the roles of CD4+ T cells and transforming growth factor beta (TGFß1) in the pathological process of valvular hyperblastosis and fibrosis of patients with rheumatic heart disease (RHD). A total of 151 patients were enrolled, among whom, 78 patients were with RHD, and 73 were age and gender matched RHD negative patients. Blood samples and valve specimens were collected for analysis. Pathological changes and collagen fibers contents of valves were analyzed using HE and Masson staining. Percentage of peripheral blood CD4+ T cells was tested through flow cytometry. TGFß1 level in serum were identified by ELISA. CD4+ T cells infiltration and expression of TGFß1, p-p38, p-JNK, p-ERK in valves were detected by immunohistochemistry. The mRNA and protein levels of p38, JNK, ERK, TGFß1, I-collagen and α-SMA were detected by qRT-PCR and western blotting, respectively. The heart valve tissues of RHD patients showed higher degrees of fibrosis, calcification and lymphocytes infiltration, which were mainly CD4+ T cells. In addition, compared with control group, RHD patients had more total CD4+ T cells in peripheral blood and valve tissues. Expression of TGFß1, phosphorylation of JNK and p38, and synthesis of I-collagen in valve tissues of RHD patients were also significantly increased. Furthermore, we found a strong positive correlation between TGFß1 expression and phosphorylation of JNK and p38. CD4+ T cells, and fibrogenic cytokine TGFß1, which activate the intracellular MAPK signaling pathway may participate in the fibrosis of heart valve in RHD patients.


Asunto(s)
Enfermedades de las Válvulas Cardíacas/genética , Estenosis de la Válvula Mitral/genética , Cardiopatía Reumática/genética , Factor de Crecimiento Transformador beta1/genética , Adulto , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/fisiología , Quinasas MAP Reguladas por Señal Extracelular/sangre , Quinasas MAP Reguladas por Señal Extracelular/genética , Femenino , Fibrosis/sangre , Fibrosis/genética , Fibrosis/patología , Regulación de la Expresión Génica/genética , Enfermedades de las Válvulas Cardíacas/sangre , Enfermedades de las Válvulas Cardíacas/patología , Humanos , MAP Quinasa Quinasa 4/sangre , MAP Quinasa Quinasa 4/genética , Sistema de Señalización de MAP Quinasas/genética , Masculino , Persona de Mediana Edad , Estenosis de la Válvula Mitral/sangre , Estenosis de la Válvula Mitral/patología , Cardiopatía Reumática/sangre , Cardiopatía Reumática/patología , Factor de Crecimiento Transformador beta1/sangre , Proteínas Quinasas p38 Activadas por Mitógenos/sangre , Proteínas Quinasas p38 Activadas por Mitógenos/genética
5.
J Cardiovasc Pharmacol ; 75(2): 141-147, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31789884

RESUMEN

Atrial apoptosis has been found to be majorly involved in the pathogenesis of human atrial fibrillation (AF). Mesencephalic astrocyte-derived neurotrophic factor exerts an antiapoptotic effect for multiple cell types. However, the correlation between MANF and atrial apoptosis in AF is still undefined. In this study, 59 patients with valvular or congenital heart disease were divided into 2 groups: AF group and sinus rhythm (SR) group. We found that the apoptotic atrial myocytes in the right atrial appendage tissues of the AF group were significantly more than those of the SR group, whereas mRNA and protein levels of MANF in the AF group were significantly down-regulated compared with those in the SR group. The serum MANF in patients with AF was markedly lower than that in patients with SR, which was inversely correlated with atrial apoptosis in patients with AF. In addition, the AF group had the greater inflammation and endoplasmic reticulum stress compared with the SR group. These findings suggest that MANF downregulation may lead to more atrial apoptosis in human chronic AF, indicating MANF as a potential therapeutic agent in AF treatment.


Asunto(s)
Apéndice Atrial/metabolismo , Fibrilación Atrial/sangre , Factores de Crecimiento Nervioso/sangre , Adulto , Anciano , Apoptosis , Apéndice Atrial/patología , Apéndice Atrial/fisiopatología , Fibrilación Atrial/diagnóstico , Fibrilación Atrial/fisiopatología , Función del Atrio Derecho , Remodelación Atrial , Biomarcadores/sangre , Estudios de Casos y Controles , Enfermedad Crónica , Regulación hacia Abajo , Estrés del Retículo Endoplásmico , Femenino , Frecuencia Cardíaca , Humanos , Mediadores de Inflamación/metabolismo , Masculino , Persona de Mediana Edad
6.
Nanotechnology ; 30(6): 065102, 2019 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-30543196

RESUMEN

Transition metal dichalogenides (TMDCs) with unique layered structures hold promising potential as transducers for photothermal therapy. However, the low photothermal conversion efficiency and poor stability in some cases limit their practical applications. Herein, we demonstrate the fabrication of ultrathin homogeneous hybridized TMDC nanosheets and their use for highly efficient photothermal tumor ablation. In particular, the nanosheets were composed of metallic WSe2 intercalated with polyvinylpyrrolidone (PVP), which was facilely prepared through a solvothermal process from the mixture of selenourea crystals, WCl6 powder along with PVP polymeric nanogel. Our characterizations revealed that the obtained nanosheets exhibited excellent photothermal conversion efficiency, therapeutic demonstration with improved biocompatibility and physiological stability attributing to the combined merits of metallic phase of WSe2 and hydrophilic PVP insertion. Both the histological analysis of vital organs and in vitro/in vivo tests confirmed the nanosheets as actively effective and biologically safe in this phototherapeutic technique. Findings from this non-invasive experiment clearly emphasize the explorable therapeutic efficacy of the layered-based hybrid agents in future cancer treatment planning procedures.


Asunto(s)
Fármacos Fotosensibilizantes/uso terapéutico , Fototerapia/métodos , Povidona/química , Selenio/química , Tungsteno/química , Animales , Línea Celular Tumoral , Femenino , Rayos Infrarrojos/uso terapéutico , Ratones , Ratones Endogámicos BALB C , Nanopartículas/química , Neoplasias Experimentales/terapia , Fármacos Fotosensibilizantes/síntesis química , Fármacos Fotosensibilizantes/química , Temperatura , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Cell Physiol Biochem ; 45(3): 1108-1120, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29439254

RESUMEN

BACKGROUND/AIMS: Qing Dai is a prized traditional Chinese medicine whose major component, indirubin, and its derivative, indirubin-3'-monoxime (IDM), have inhibitory effects on the growth of many human tumor cells and pronounced anti-leukemic activities. However, the effects of IDM on mature human erythrocytes are unclear. This study aimed to evaluate the potential impact of IDM on erythrocytes and the mechanisms underlying that impact. METHODS: Utilizing flow cytometry and confocal laser scanning microscopy, phosphatidylserine exposure at the cell surface was estimated by annexin V-fluorescein isothiocyanate (FITC). The relative cell size, expressed in arbitrary units, was evaluated by forward scatter in a flow cytometer. Fluo-3 fluorescence was used to bewrite changes in cytosolic Ca2+ activity, reactive oxygen species (ROS) formation was assessed by 2,7-dichlorodihydrofluorescein diacetate (DCFH-DA) fluorescence, and ceramide abundance was evaluated by FITC-conjugated specific antibodies. RESULTS: The 24-h exposure of human erythrocytes to IDM (12 µM) significantly decreased the percentage of annexin V-binding erythrocytes and the intracellular calcium concentration ([Ca2+]i). IDM (3-12 µM) did not significantly modify the ceramide level or DCFH-DA fluorescence. Energy depletion (removal of glucose for 24 hours) significantly increased annexin V binding and Fluo-3 fluorescence and diminished forward scatter, and these effects were significantly mitigated by IDM (12 µM). Moreover, the Ca2+ ionophore ionomycin (1 µM, 60 min) and oxidative stress (30 min exposure to 0.05 mM tert-butyl hydroperoxide, t-BHP) similarly triggered eryptosis, which was also significantly suppressed by IDM. CONCLUSIONS: IDM is a novel inhibitor of suicidal erythrocyte death following ionomycin treatment, t-BHP treatment and energy depletion. Thus, IDM may counteract anemia and impairment of microcirculation, at least in part, by inhibition of Ca2+ entry into erythrocytes.


Asunto(s)
Eriptosis/efectos de los fármacos , Indoles/farmacología , Oximas/farmacología , Compuestos de Anilina/química , Calcio/metabolismo , Tamaño de la Célula/efectos de los fármacos , Células Cultivadas , Ceramidas/metabolismo , Membrana Eritrocítica/efectos de los fármacos , Eritrocitos/citología , Eritrocitos/efectos de los fármacos , Eritrocitos/metabolismo , Citometría de Flujo , Humanos , Ionomicina/farmacología , Medicina Tradicional China , Microscopía Confocal , Estrés Oxidativo/efectos de los fármacos , Fosfatidilserinas/farmacología , Especies Reactivas de Oxígeno/metabolismo , Xantenos/química , terc-Butilhidroperóxido/farmacología
8.
J Neuroinflammation ; 15(1): 152, 2018 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-29776417

RESUMEN

BACKGROUND: Epilepsy is a common neurological disorder and is not well controlled by available antiepileptic drugs (AEDs). Inflammation is considered to be a critical factor in the pathophysiology of epilepsy. Sinomenine (SN), a bioactive alkaloid with anti-inflammatory effect, exerts neuroprotective activity in many nervous system diseases. However, little is known about the effect of SN on epilepsy. METHODS: The chronic epilepsy model was established by pentylenetetrazole (PTZ) kindling. Morris water maze (MWM) was used to test spatial learning and memory ability. H.E. staining and Hoechst 33258 staining were used to evaluate hippocampal neuronal damage. The expression of nucleotide oligomerization domain (NOD)-like receptor protein 1 (NLRP1) inflammasome complexes and the level of inflammatory cytokines were determined by western blot, quantitative real-time PCR and enzyme-linked immunosorbent assay (ELISA) kits. RESULTS: SN (20, 40, and 80 mg/kg) dose-dependently disrupts the kindling acquisition process, which decreases the seizure scores and the incidence of fully kindling. SN also increases the latency of seizure and decreases the duration of seizure in fully kindled rats. In addition, different doses of SN block the hippocampal neuronal damage and minimize the impairment of spatial learning and memory in PTZ kindled rats. Finally, PTZ kindling increases the expression of NLRP1 inflammasome complexes and the levels of inflammatory cytokines IL-1ß, IL-18, IL-6, and TNF-α, which are all attenuated by SN in a dose- dependent manner. CONCLUSIONS: SN exerts anticonvulsant and neuroprotective activity in PTZ kindling model of epilepsy. Disrupting the kindling acquisition, which inhibits NLRP1 inflammasome-mediated inflammatory process, might be involved in its effects.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Epilepsia/tratamiento farmacológico , Excitación Neurológica/efectos de los fármacos , Morfinanos/uso terapéutico , Proteínas del Tejido Nervioso/metabolismo , Análisis de Varianza , Animales , Proteínas Adaptadoras de Señalización CARD/genética , Proteínas Adaptadoras de Señalización CARD/metabolismo , Caspasa 1/genética , Caspasa 1/metabolismo , Convulsivantes/toxicidad , Citocinas/metabolismo , Modelos Animales de Enfermedad , Epilepsia/inducido químicamente , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Proteínas del Tejido Nervioso/genética , Pentilenotetrazol/toxicidad , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley
9.
Brain Behav Immun ; 69: 255-263, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29195783

RESUMEN

Blood-brain barrier (BBB) disruption, thrombus formation and immune-mediated inflammation are important steps in the pathophysiology of cerebral ischemia-reperfusion injury but are still inaccessible to therapeutic interventions. Recent studies have provided increasing evidence that blocking of platelet glycoprotein (GP) receptor Ib might represent a novel target in treating acute ischemic stroke. This research was conducted to explore the therapeutic efficacy and potential mechanisms of GPIbα inhibitor (anfibatide) in a model of brain ischemia-reperfusion injury in mice. Male mice underwent 90 min of right middle cerebral artery occlusion (MCAO) followed by 24 h of reperfusion. Anfibatide (1, 2, 4 ug/kg) or tirofiban were administered intravenously 1 h after reperfusion. The results showed that anfibatide could significantly reduce infarct volumes, increase the number of intact neuronal cells and improve neurobehavioral function. Moreover, anfibatide could reduce post ischemic BBB damage by attenuating increased paracellular permeability in the ischemia hemisphere significantly. Stroke-induced increases in activity and protein expression of macrophage-1 antigen (MAC-1) and P-selectin were also reduced by anfibatide intervention. Finally, anfibatide exerted antithrombotic effects upon stroke by decreased the number of microthrombi formation. This is the first demonstration of anfibatide's efficacy in protecting the BBB integrity and decreasing neutrophil inflammation response mediated by MAC-1 besides microthrombus formation inhibition in the brain during reperfusion. Anfibatide, as a promising anti-thrombo-inflammation agent, could be beneficial for the treatment of ischemic stroke.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Venenos de Crotálidos/uso terapéutico , Fibrinolíticos/uso terapéutico , Lectinas Tipo C/uso terapéutico , Complejo GPIb-IX de Glicoproteína Plaquetaria/antagonistas & inhibidores , Daño por Reperfusión/tratamiento farmacológico , Tirofibán/uso terapéutico , Animales , Barrera Hematoencefálica/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Venenos de Crotálidos/farmacología , Fibrinolíticos/farmacología , Infarto de la Arteria Cerebral Media/metabolismo , Masculino , Ratones , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Daño por Reperfusión/metabolismo , Tirofibán/farmacología
10.
J Neuroinflammation ; 14(1): 139, 2017 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-28732502

RESUMEN

BACKGROUND: Neuroinflammation mediated by NLRP1 (nucleotide-binding oligomerization domain (NOD)-like receptor protein 1) inflammasome plays an important role in many neurological diseases such as Parkinson's disease (PD) and Alzheimer's disease (AD). Our previous studies showed that chronic glucocorticoid (GC) exposure increased brain inflammation via NLRP1 inflammasome and induce neurodegeneration. However, little is known about the mechanism of chronic GC exposure on NLRP1 inflammasome activation in hippocampal neurons. METHODS: Hippocampal neurons damage was assessed by LDH kit and Hoechst 33258 staining. The expression of microtubule-associated protein 2 (MAP2), inflammasome complex protein (NLRP1, ASC and caspase-1), inflammatory cytokines (IL-1ß), and large-conductance Ca2+ and voltage-activated K+ channel (BK channels) protein was detected by Western blot. The inflammatory cytokines (IL-1ß and IL-18) were examined by ELISA kit. The mRNA levels of NLRP1, IL-1ß, and BK were detected by real-time PCR. BK channel currents were recorded by whole-cell patch-clamp technology. Measurement of [K+]i was performed by ion-selective electrode (ISE) technology. RESULTS: Chronic dexamethasone (DEX) treatment significantly increased LDH release and neuronal apoptosis and decreased expression of MAP2. The mechanistic studies revealed that chronic DEX exposure significantly increased the expression of NLRP1, ASC, caspase-1, IL-1ß, L-18, and BK protein and NLRP1, IL-1ß and BK mRNA levels in hippocampal neurons. Further studies showed that DEX exposure results in the increase of BK channel currents, with the subsequent K+ efflux and a low concentration of intracellular K+, which involved in activation of NLRP1 inflammasome. Moreover, these effects of chronic DEX exposure could be blocked by specific BK channel inhibitor iberiotoxin (IbTx). CONCLUSION: Our findings suggest that chronic GC exposure may increase neuroinflammation via activation of BK-NLRP1 signal pathway and promote hippocampal neurons damage, which may be involved in the development and progression of AD.


Asunto(s)
Lesiones Encefálicas/inducido químicamente , Dexametasona/toxicidad , Hipocampo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Sinaptotagminas/metabolismo , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Lesiones Encefálicas/patología , Células Cultivadas , Modelos Animales de Enfermedad , Glucocorticoides/toxicidad , Hipocampo/efectos de los fármacos , Hipocampo/patología , L-Lactato Deshidrogenasa/metabolismo , Potenciales de la Membrana/efectos de los fármacos , Ratones , Ratones Endogámicos ICR , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas del Tejido Nervioso/genética , Péptidos/toxicidad , Ratas , Sinaptotagminas/genética
11.
Tumour Biol ; 39(6): 1010428317705750, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28618956

RESUMEN

Acid-sensing ion channels, a proton-gated cation channel, can be activated by low extracellular pH and involved in pathogenesis of some tumors such as glioma and breast cancer. However, the role of acid-sensing ion channels in the growth of lung cancer cell is unclear. In this study, we investigated the expression of acid-sensing ion channels in human lung cancer cell line A549 and their possible role in proliferation and migration of A549 cells. The results show that acid-sensing ion channel 1, acid-sensing ion channel 2, and acid-sensing ion channel 3 are expressed in A549 cells at the messenger RNA and protein levels, and acid-sensing ion channel-like currents were elicited by extracellular acid stimuli. Moreover, we found that acidic extracellular medium or overexpressing acid-sensing ion channel 1a promotes proliferation and migration of A549 cells. In addition psalmotoxin 1, a specific acid-sensing ion channel 1a inhibitor, or acid-sensing ion channel 1a knockdown can abolish the effect of acid stimuli on A549 cells. In addition, acid-sensing ion channels mediate increase of [Ca2+]i induced by low extracellular pH in A549 cells. All these results indicate that acid-sensing ion channel-calcium signal mediate lung cancer cell proliferation and migration induced by extracellular acidosis, and acid-sensing ion channels may serve as a prognostic marker and a therapeutic target for lung cancer.


Asunto(s)
Canales Iónicos Sensibles al Ácido/biosíntesis , Biomarcadores de Tumor/biosíntesis , Neoplasias Pulmonares/genética , Células A549 , Canales Iónicos Sensibles al Ácido/genética , Acidosis/genética , Acidosis/patología , Movimiento Celular/genética , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Neoplasias Pulmonares/patología , Pronóstico
13.
Cell Physiol Biochem ; 40(6): 1391-1400, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27997909

RESUMEN

BACKGROUND: The progression of systemic lupus erythematosus (SLE) leads to anemia in patients, adversely affecting prognosis. The diverse causes of anemia may include excessive eryptosis or premature suicidal erythrocyte death characterized by cell shrinkage and phosphatidylserine (PS) exposure on the cell surface. The present study explored if SLE enhances eryptosis and the underlying mechanisms. MATERIALS AND METHODS: Eryptosis was assessed using flow cytometry in healthy volunteers (n = 20) and anemic patients hospitalized for SLE (n = 22), for parameters including PS exposure, cell volume, cytosolic calcium ion (Ca2+) levels and reactive oxygen species (ROS) and ceramide abundance. These indicators were measured in erythrocytes of experimental subjects and erythrocytes treated with plasma from healthy volunteers or SLE patients. RESULTS: The hemoglobin and hematocrit levels were significantly lower in anemic SLE patients than in healthy volunteers (***p<0.001, p<0.001, respectively). The percentage of PS-exposing erythrocytes was significantly higher in SLE patients than in healthy volunteers (p<0.001), accompanied by an increase in cytosolic Ca2+ levels, oxidative stress. The measurements of PS and Ca2+ levels were significantly higher in the erythrocytes of healthy volunteers following incubation in plasma of SLE patients than in plasma of healthy volunteers for 24h (***p<0.001, *p<0.05 respectively). CONCLUSION: Eryptosis is enhanced in SLE and may contribute to anemia. The probable underlying mechanisms may be an excessive formation of ROS in erythrocytes. Also, some plasma components may trigger eryptosis by increasing the cytosolic Ca2+ concentration.


Asunto(s)
Anemia/complicaciones , Anemia/patología , Eriptosis , Lupus Eritematoso Sistémico/complicaciones , Lupus Eritematoso Sistémico/patología , Adolescente , Adulto , Anciano , Anemia/sangre , Antioxidantes/metabolismo , Calcio/metabolismo , Ceramidas/metabolismo , Citosol/metabolismo , Eritrocitos/metabolismo , Femenino , Voluntarios Sanos , Humanos , Lupus Eritematoso Sistémico/sangre , Masculino , Persona de Mediana Edad , Fosfatidilserinas/metabolismo , Plasma/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Adulto Joven
14.
Int Immunol ; 27(7): 317-25, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25840437

RESUMEN

Accumulating evidence has identified the profibrogenic properties of IL-17A in organ fibrosis. However, the role of IL-17A signal in liver fibrosis induced by Schistosoma japonicum infection remains unclear. In this study, we investigated liver fibrosis in wild-type (WT) and IL-17RA(-/-) mice upon S. japonicum infection. Hepatic IL-17A, IL-17C, IL-17E (IL-25), IL-17F, IL-17RA, IL-17RB and IL-17RC transcript levels were determined by RT-PCR. IL-17A(+) cells were analyzed by flow cytometry and confocal microscopy among granuloma cells. Immunostaining of IL-17R was performed on liver sections. Collagen deposition was assessed by Van Gieson's staining. IL-17A, IL-17C, IL-17E, IL-17F, IL-17RA and IL-17RC mRNA levels were dramatically increased in fibrotic livers. Among granuloma cells, CD3(+) and CD3(-) lymphocytes, neutrophils and macrophages were found to express IL-17A. Compared to WT, IL-17RA(-/-) mice displayed attenuated granulomatous inflammation, liver fibrosis, improved liver function and high survival. Meanwhile, α-smooth muscle actin staining and the expression of fibrogenic genes (transforming growth factor ß, IL-13 and collagen-I) as well as IL-17A-induced proinflammatory mediators (IL-1ß, IL-6, tumor necrosis factor α, CXCL1 and CXCL2) and proteinases (MMP3 and TIMP1) involved in fibrosis were markedly reduced in IL-17RA(-/-) mice. In addition, Th2 cytokines IL-4 and IL-17E (IL-25) were also decreased in IL-17RA(-/-) mice. These results indicated that IL-17A signal contributes to the pathogenesis of liver fibrosis in murine schistosomiasis. This effect was induced possibly by activating hepatic stellate cells and stimulating the release of proinflammatory cytokines and chemokines. Furthermore, the Th2 response was also enhanced by IL-17A signals. Our data demonstrate that IL-17A may serve as a promising target for antifibrotic therapy.


Asunto(s)
Interleucina-17/inmunología , Cirrosis Hepática/inmunología , Esquistosomiasis Japónica/inmunología , Transducción de Señal/inmunología , Animales , Femenino , Interleucina-17/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
15.
Brain Behav Immun ; 52: 58-70, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26434621

RESUMEN

Neuroinflammation plays an important role in the pathogenesis of neurodegenerative diseases, such as Alzheimer's disease (AD) and depression. Chronic glucocorticoids (GCs) exposure has deleterious effects on the structure and function of neurons and is associated with development and progression of AD. However, little is known about the proinflammatory effects of chronic GCs exposure on neurodegeneration in brain. Therefore, the aim of this study was to evaluate the effects of chronic dexamethasone (DEX) treatment (5mg/kg, s.c. for 7, 14, 21 and 28 days) on behavior, neurodegeneration and neuroinflammatory parameters of nucleotide-binding oligomerization domain-like receptor pyrin domain-containing 1 (NLRP-1) inflammasome in male mice. The results showed that DEX treatment for 21 and 28 days significantly reduced the spontaneous motor activity and exploratory behavior of the mice. In addition, these mice showed significant neurodegeneration and a decrease of microtubule-associated protein 2 (MAP2) in the frontal cortex and hippocampus CA3. DEX treatment for 7, 14, 21 and 28 days significantly decreased the mRNA and protein expression of glucocorticoid receptor (GR). Moreover, DEX treatment for 21 and 28 days significantly increased the proteins expression of NLRP-1, Caspase-1, Caspase-5, apoptosis associated speck-like protein (ASC), nuclear factor-κB (NF-κB), p-NF-κB, interleukin-1ß (IL-1ß), IL-18 and IL-6 in the frontal cortex and hippocampus brain tissue. DEX treatment for 28 days also significantly increased the mRNA expression levels of NLRP-1, Caspase-1, ASC and IL-1ß. These results suggest that chronic GCs exposure may increase brain inflammation via NLRP-1 inflammasome activation and induce neurodegeneration.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Dexametasona/administración & dosificación , Lóbulo Frontal/efectos de los fármacos , Hipocampo/efectos de los fármacos , Inflamasomas/efectos de los fármacos , Animales , Caspasa 1/metabolismo , Proteínas del Citoesqueleto/metabolismo , Dexametasona/metabolismo , Lóbulo Frontal/metabolismo , Lóbulo Frontal/patología , Hipocampo/metabolismo , Hipocampo/patología , Inflamasomas/metabolismo , Interleucinas/metabolismo , Masculino , Ratones , Ratones Endogámicos ICR , FN-kappa B/metabolismo , Distribución Aleatoria , Receptores de Glucocorticoides/metabolismo
16.
Mol Carcinog ; 54(10): 1086-95, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25043657

RESUMEN

Recently, an increasing number of studies have suggested that mTOR plays a critical role in the regulation of tumor cell motility, invasion and cancer metastasis. However, little is known about the signaling mechanisms in regulating epithelial-mesenchymal transition (EMT) of prostate cancer. In this study, we found that the expression levels of Raptor and Rictor in prostate cancer tissues were elevated, which may suggest that Raptor and Rictor signaling pathways are associated with prostate cancer progression and metastasis. Inhibition of mTORC1 or mTORC2 by knock down of Raptor or Rictor, respectively, migration and invasion of prostate cancer were attenuated. Furthermore, EMT, a characterized by the changed expression levels of various markers like E-cadherin, ß-catenin, N-cadherin, and vimentin emergend following inhibition of Raptor or Rictor. Finally, the small GTPases (RhoA and Rac1) which were crucial regulatory proteins in cell migration and invasion were inactivited after downregulating Raptor and Rictor. These results suggest that mTOR regulate EMT at least in part by down regulation of RhoA and Rac1 signaling pathways. Our findings provide novel very attractive target strategies that the inhibition of mTOR signaling pathways may retard prostate cancer migration and invasion at early stages.


Asunto(s)
Transición Epitelial-Mesenquimal/genética , Neoplasias de la Próstata/genética , Transducción de Señal/genética , Serina-Treonina Quinasas TOR/genética , Proteína de Unión al GTP rac1/genética , Proteína de Unión al GTP rhoA/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Biomarcadores de Tumor/genética , Cadherinas/genética , Proteínas Portadoras/genética , Línea Celular Tumoral , Movimiento Celular/genética , Regulación hacia Abajo/genética , Transición Epitelial-Mesenquimal/fisiología , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Masculino , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Neoplasias de la Próstata/patología , Proteína Asociada al mTOR Insensible a la Rapamicina , Proteína Reguladora Asociada a mTOR , Vimentina/genética , beta Catenina/genética
17.
J Comp Neurol ; 532(6): e25629, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-39031887

RESUMEN

In the brain, connectivity determines function. Neurons in the parabrachial nucleus (PB) relay diverse information to widespread brain regions, but the connections and functions of PB neurons that express Nps (neuropeptide S, NPS) remain mysterious. Here, we use Cre-dependent anterograde tracing and whole-brain analysis to map their output connections. While many other PB neurons project ascending axons through the central tegmental tract, NPS axons reach the forebrain via distinct periventricular and ventral pathways. Along the periventricular pathway, NPS axons target the tectal longitudinal column and periaqueductal gray, then continue rostrally to target the paraventricular nucleus of the thalamus. Along the ventral pathway, NPS axons blanket much of the hypothalamus but avoid the ventromedial and mammillary nuclei. They also project prominently to the ventral bed nucleus of the stria terminalis, A13 cell group, and magnocellular subparafasciular nucleus. In the hindbrain, NPS axons have fewer descending projections, targeting primarily the superior salivatory nucleus, nucleus of the lateral lemniscus, and periolivary region. Combined with what is known already about NPS and its receptor, the output pattern of Nps-expressing neurons in the PB region predicts roles in threat response and circadian behavior.


Asunto(s)
Núcleos Parabraquiales , Animales , Núcleos Parabraquiales/fisiología , Núcleos Parabraquiales/citología , Ratones , Vías Eferentes/citología , Vías Eferentes/fisiología , Ratones Transgénicos , Neuronas/metabolismo , Masculino , Neuropéptidos/metabolismo , Vías Nerviosas/citología
18.
Immunobiology ; 229(1): 152778, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38159526

RESUMEN

Mesencephalic astrocyte-derived neurotrophic factor (MANF) is an endoplasmic reticulum (ER) stress-induced protein, and it has been reported that ER stress and unfolded protein response (UPR) are closely related to the immune system. The spleen is an important immune organ and we have shown in our previous research that MANF is expressed in human spleen tissues. However, there have been limited studies about the effect of MANF on spleen development. In this study, we detected MANF expression in spleen tissues and found that MANF was expressed in the red pulp and marginal zone. Additionally, MANF was localized in the CD68+ and CD138+ cells of adult rat spleen tissues, but not in the CD3+ cells. We performed immunohistochemical staining to detect MANF expression in the spleen tissues of rats that were different ages, and we found that MANF+ cells were localized together in the spleen tissues of rats that were 1-4 weeks old. MANF was also expressed in CD68+ cells in the spleen tissues of rats and mice. Furthermore, we found that MANF deficiency inhibited white pulp development in MANF knockout mice, thus indicating that MANF played an important role in the white pulp development of rodent spleen tissues.


Asunto(s)
Astrocitos , Bazo , Animales , Humanos , Ratones , Ratas , Astrocitos/metabolismo , Estrés del Retículo Endoplásmico , Ratones Noqueados , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/metabolismo , Factores de Crecimiento Nervioso/farmacología , Bazo/metabolismo , Respuesta de Proteína Desplegada
19.
Phytomedicine ; 129: 155661, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38677269

RESUMEN

BACKGROUND: Gallbladder cancer (GBC) poses a significant risk to human health. Its development is influenced by numerous factors, particularly the homeostasis of reactive oxygen species (ROS) within cells. This homeostasis is crucial for tumor cell survival, and abnormal regulation of ROS is associated with the occurrence and progression of many cancers. Dihydrotanshinone I (DHT I), a biologically effective ingredient isolated from Salvia miltiorrhiza, has exhibited cytotoxic properties against various tumor cells by inducing apoptosis. However, the precise molecular mechanisms by which dht I exerts its cytotoxic effects remain unclear. PURPOSE: To explore the anti-tumor impact of dht I on GBC and elucidate the potential molecular mechanisms. METHODS: The proliferation of GBC cells, NOZ and SGC-996, was assessed using various assays, including CCK-8 assay, colony formation assay and EdU staining. We also examined cell apoptosis, cell cycle progression, ROS levels, and alterations in mitochondrial membrane potential to delve into the intricate molecular mechanism. Quantitative PCR (qPCR), immunofluorescence staining, and Western blotting were performed to evaluate target gene expression at both the mRNA and protein levels. The correlation between nuclear factor erythroid 2-related factor 2 (Nrf2) and kelch-like ECH-associated protein 1 (Keap1) were examined using co-immunoprecipitation. Finally, the in vivo effect of dht I was investigated using a xenograft model of gallbladder cancer in mice. RESULTS: Our research findings indicated that dht I exerted cytotoxic effects on GBC cells, including inhibiting proliferation, disrupting mitochondrial membrane potential, inducing oxidative stress and apoptosis. Our in vivo studies substantiated the inhibition of dht I on tumor growth in xenograft nude mice. Mechanistically, dht I primarily targeted Nrf2 by promoting Keap1 mediated Nrf2 degradation and inhibiting protein kinase C (PKC) induced Nrf2 phosphorylation. This leads to the suppression of Nrf2 nuclear translocation and reduction of its target gene expression. Moreover, Nrf2 overexpression effectively counteracted the anti-tumor effects of dht I, while Nrf2 knockdown significantly enhanced the inhibitory effect of dht I on GBC. Meanwhile, PKC inhibitors and nuclear import inhibitors increased the sensitivity of GBC cells to dht I treatment. Conversely, Nrf2 activators, proteasome inhibitors, antioxidants and PKC activators all antagonized dht I induced apoptosis and ROS generation in NOZ and SGC-996 cells. CONCLUSION: Our findings indicated that dht I inhibited the growth of GBC cells by regulating the Keap1-Nrf2 signaling pathway and Nrf2 phosphorylation. These insights provide a strong rationale for further investigation of dht I as a potential therapeutic agent for GBC treatment.


Asunto(s)
Apoptosis , Proliferación Celular , Neoplasias de la Vesícula Biliar , Proteína 1 Asociada A ECH Tipo Kelch , Ratones Desnudos , Factor 2 Relacionado con NF-E2 , Fenantrenos , Especies Reactivas de Oxígeno , Transducción de Señal , Animales , Humanos , Ratones , Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Furanos/farmacología , Neoplasias de la Vesícula Biliar/tratamiento farmacológico , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones Endogámicos BALB C , Factor 2 Relacionado con NF-E2/metabolismo , Fenantrenos/farmacología , Fosforilación/efectos de los fármacos , Quinonas/farmacología , Especies Reactivas de Oxígeno/metabolismo , Salvia miltiorrhiza/química , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Heliyon ; 10(5): e27618, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38495177

RESUMEN

Acyl-CoA thioesterase 4 (ACOT4) has been reported to be related to acetyl-CoA carboxylase activity regulation; However, its exact functions in liver lipid and glucose metabolism are still unclear. Here, we discovered explored the regulatory roles of ACOT4 in hepatic lipid and glucose metabolism in vitro. We found that the expression level of ACOT4 was significantly increased in the hepatic of db/db and ob/ob mice as well as obese mice fed a high fat diet. Adenovirus-mediated overexpression of ACOT4 promoted gluconeogenesis and high-glucose/high-insulin-induced lipid accumulation and impaired insulin sensitivity in primary mouse hepatocytes, whereas ACOT4 knockdown notably suppressed gluconeogenesis and decreased the triglycerides accumulation in hepatocytes. Furthermore, ACOT4 knockdown increased insulin-induced phosphorylation of AKT and GSK-3ß in primary mouse hepatocytes. Mechanistically, we found that upregulation of ACOT4 expression inhibited AMP-activated protein kinase (AMPK) activity, and its knockdown had the opposite effect. However, activator A769662 and inhibitor compound C of AMPK suppressed the impact of the change in ACOT4 expression on AMPK activity. Our data indicated that ACOT4 is related to hepatic glucose and lipid metabolism, primarily via the regulation of AMPK activity. In conclusion, ACOT4 is a potential target for the therapy of non-alcoholic fatty liver (NAFLD) and type 2 diabetes.

SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda